bims-imseme Biomed News
on Immunosenescence and T cell metabolism
Issue of 2025–09–28
thirty-one papers selected by
Pierpaolo Ginefra, Ludwig Institute for Cancer Research



  1. Trends Endocrinol Metab. 2025 Sep 22. pii: S1043-2760(25)00190-0. [Epub ahead of print]
      CD8+ T cells are vital for antiviral and antitumor immunity, yet in hostile microenvironments, they experience metabolic stress, leading to mitochondrial damage, metabolic dysregulation, and chromatin remodeling that cause immune dysfunction. Aging further exacerbates these processes, with intrinsic metabolic collapse and extrinsic environmental factors jointly impairing T cell immunity. Metabolites orchestrate key epigenetic modifications, shaping transcriptional programs essential for T cell differentiation and memory formation. This review explores the interconnected metabolic and epigenetic mechanisms governing CD8+ T cell fate decisions, emphasizing how mitochondrial dysfunction, metabolic inflexibility, and nutrient competition drive CD8+ T cell exhaustion, senescence, and age-associated dysfunction. Understanding these metabolic-epigenetic circuits offers novel therapeutic avenues, including metabolic reprogramming and senescence-targeted strategies, to rejuvenate immune responses and enhance immunotherapy outcomes.
    Keywords:  T cell; aging; exhaustion; metabolism; senescence
    DOI:  https://doi.org/10.1016/j.tem.2025.08.009
  2. Cell Rep. 2025 Sep 18. pii: S2211-1247(25)01087-3. [Epub ahead of print]44(10): 116316
      CD8+ T cell exhaustion limits immune responses during cancer and chronic infection. We identify CD7 as a tissue-specific regulator of terminally exhausted CD8+ T cells during chronic infection. CD7 expression progressively increases during exhaustion, reaching its highest levels on a subset of CD101+Tim3low terminally exhausted cells that arise in the liver. Transcriptomic analysis revealed that CD7-deficient terminally exhausted cells display altered expression of co-stimulatory, translational, and effector genes, correlating with markedly reduced persistence and increased apoptotic susceptibility. Importantly, CD7 is preferentially upregulated on PD-1+CD39+ tumor-infiltrating lymphocytes in human head and neck squamous cell carcinoma, suggesting that CD7 may play a conserved role in promoting exhausted T cell survival. These findings reveal a function for CD7 in sustaining terminally exhausted CD8+ T cells and demonstrate that CD7 signaling is a critical regulator of T cell persistence during chronic infection.
    Keywords:  CD7; CD8 T cell; CP: Immunology; LCMV; T cell; T cell exhaustion; immunology; scRNA-seq; viral immunology
    DOI:  https://doi.org/10.1016/j.celrep.2025.116316
  3. Sci Adv. 2025 Sep 26. 11(39): eadw6755
      Glycans regulate cellular function, yet how aging affects the glycocalyx remains unclear. Here, we investigate changes in immune cell glycocalyx with age and find that α2,6-linked sialic acid, a glycan epitope associated with inhibitory signaling, is down-regulated in T cells from old animals. This reduction is tightly correlated with age-associated accumulation of effector T cells, which have little to no α2,6-linked sialic acid. To understand how α2,6-linked sialic acid affects T cell physiology, we generated a mouse model with T cell-specific deletion of sialyltransferase gene St6gal1. The lack of α2,6-linked sialic acid leads to reduced responsiveness in naïve T cells, leading to impaired T cell responses against Listeria monocytogenes infection and tumor growth. PD-1 pathway blockade partially restores St6gal1-deficient T cells' ability to control tumor growth. These findings suggest that α2,6-linked sialic acid is critical for maintaining long-term T cell responsiveness, and its loss may contribute to decreased T cell function with age.
    DOI:  https://doi.org/10.1126/sciadv.adw6755
  4. Mater Today Bio. 2025 Oct;34 102258
      Bone tumors establish a self-perpetuating vicious cycle wherein metabolic reprogramming (e.g., aerobic glycolysis, glutamine addiction) drives both T cell exhaustion and osteolytic damage. Tumor-derived lactate and nutrient depletion suppress T cell function while promoting osteoclast activation and inhibiting osteoblast differentiation. Reciprocally, bone damage releases immunosuppressive factors (e.g., TGF-β, and calcium) that further exacerbate T cell exhaustion, creating a pathological feedback loop. This review proposes the "Metabolic-Immune-Bone Network" (MIBN) as a framework for understanding this interplay. Crucially, multifunctional nanomaterials offer a promising strategy to disrupt this cycle. By precisely targeting metabolic pathways, they simultaneously suppress tumor growth and alleviate microenvironmental immunosuppression/acidosis. Their multifunctional design enables co-delivery of metabolic inhibitors, immune modulators, and osteogenic agents, thereby restoring T cell cytotoxicity and promoting bone regeneration. This dual "anti-tumor and osseous-preserving" functionality addresses the limitations of conventional therapies, shifting the paradigm from lesion-focused treatment toward holistic rehabilitation. This aligns with the "3R" strategy-Remodel, Repair, and Remove-highlighting microenvironment modulation, bone regeneration, and immune-mediated tumor clearance. Future advances in stimulus-responsive and metabolically targeted nanomaterials hold significant potential for breaking the MIBN-driven vicious cycle in bone oncology.
    Keywords:  Aerobic glycolysis; Bone Tumor microenvironment; Bone damage; Glutamine metabolism; Metabolic reprogramming; Nanomaterials; T cell exhaustion
    DOI:  https://doi.org/10.1016/j.mtbio.2025.102258
  5. Int J Mol Sci. 2025 Sep 10. pii: 8799. [Epub ahead of print]26(18):
      Recently, senescent T cells in the peripheral blood have been detected using senescence-associated β-galactosidase (SA-βGal) activity and have been used as an endpoint in clinical trials. However, the epidemiological association between the abundance of SA-βGal-stained senescent CD8+ T cells and chronological age has not been fully elucidated. To examine the correlation between the proportion of SA-βGalhigh CD8+ T cells and age, we analyzed previously collected clinical trial data. We conducted a cross-sectional analysis of 632 Japanese adults aged 40-59 years who participated in the screening phase of a clinical trial. To characterize senescent CD8+ T cells, we measured the proportion of SA-βGalhigh in total CD8+ T cells and each subset-naïve, central memory (TCM), effector memory (TEM), and terminally differentiated effector memory (TEMRA). We then calculated the correlation coefficients between the proportion of SA-βGalhigh CD8+ T cells and age. The proportion of SA-βGalhigh cells in total CD8+ T cells, naïve, TCM, TEM, and TEMRA CD8+ T cells increased significantly with age. In Japanese adults, the proportion of SA-βGalhigh in CD8+ T cells may serve as a useful biomarker of immune senescence.
    Keywords:  CD8+ T cells; age; senescence; senescence-associated β-galactosidase
    DOI:  https://doi.org/10.3390/ijms26188799
  6. Nat Aging. 2025 Sep 25.
      Age-related increases in cancer have traditionally been attributed to compromised antitumor immunity of exhausted and dysfunctional CD8⁺ T cells. Here we provide an alternative mechanism: in aging, cancer also progresses with the help of fully functional CD8⁺ T cells. These transcriptionally and epigenetically distinct cells (termed double-positive CD8+ T cells (DP8)) express CD39, CD73, CD101 and CXCR6 on their surface and accumulate during healthy aging in mice, requiring B cells presenting cognate antigens. In aged mice, progressing tumors recruit DP8 cells via the CXCL16-CXCR6 axis to suppress antitumor CD4+ T cells in an ADP/adenosine-dependent manner, and targeting DP8 cell function or recruitment can reverse tumor growth in aged mice. This tumor-promoting mechanism of DP8 cells appears to be conserved in older humans, as we detected DP8-like cells in various tumors, including late-onset breast cancer. We propose that this tumor-promoting role of CD8+ T cells should be considered in the development of therapeutics tailored for older humans.
    DOI:  https://doi.org/10.1038/s43587-025-00966-3
  7. Nature. 2025 Sep 26.
      
    Keywords:  Ageing; Cell biology; Genetics; Metabolism
    DOI:  https://doi.org/10.1038/d41586-025-03064-x
  8. bioRxiv. 2025 Sep 20. pii: 2025.09.18.677001. [Epub ahead of print]
      We and others have reported evidence of T cell exhaustion in children with perinatal HIV with increased expression of inhibitory receptors PD-1, CD160, and TIM-3, but there is limited data on the virologic functional consequences of this immune exhaustion. We address this by using an immune database from Kenyan children with perinatal HIV and unexposed controls. We computationally integrate T cell profiles of differentiation, activation and exhaustion in an agent-based model (ABM) to predict how T cell exhaustion impacts viral control following HIV exposure in vitro. Our ABM includes macrophages, CD4 and CD8 T cells, cytokines, and HIV. Model mechanisms include viral dynamics, macrophage activation, T cell activation and proliferation, cytotoxic T cell killing, and cytokine/HIV diffusion and degradation. Participants are grouped by HIV plasma viremia and by age, less than 5 years or 5-18 years. Our findings indicate that cells from virally active participants, who have the highest levels of exhaustion, have lower predicted viral concentrations and infected cells compared to other participant groups during new infection. However, this coincides with higher cell death, suggesting that short-term viral control is associated with excessive inflammation, which could be detrimental long-term. Cells from virally suppressed participants older than 5 years can maintain lower viral concentrations while limiting cell death, reflecting a more sustainable short-term immune response. In virally suppressed children younger than 5 years, immune response patterns strongly resemble the age-matched healthy control group, suggesting early viral suppression may preserve antiviral immune responses. Our model predicts unique patterns of cell death for each participant group, with CD8 T cell death being dominant in virally active groups and CD4 T cell and macrophage death being dominant in healthy and virally suppressed groups. Finally, exhausted CD8 T cells are predicted to contribute significantly to CD8 T cell killing, proliferation, and activation in the virally active group, indicating partially functional CD8 T cells can still contribute to short-term viral control. Our analysis functionally integrates participant-specific immunophenotypic data to allow quantification of the extent, mechanisms, and impact of immune dysfunction in perinatal HIV and could inform pediatric HIV remission and cure strategies.
    DOI:  https://doi.org/10.1101/2025.09.18.677001
  9. Crit Rev Oncol Hematol. 2025 Sep 24. pii: S1040-8428(25)00356-7. [Epub ahead of print] 104968
      Chimeric Antigen Receptor T cell (CAR-T) therapy has revolutionized cancer treatment, achieving remarkable success in hematological malignancies. However, its efficacy against solid tumors remains limited, primarily due to challenges such as the immunosuppressive tumor microenvironment (TME) and T cell exhaustion. Protein post-translational modifications (PTMs), including phosphorylation, ubiquitination, glycosylation, acetylation, and lactylation, are pivotal in regulating T cell signaling, activation, persistence, and metabolic adaptation, thereby offering novel opportunities to enhance CAR-T therapy. The impact of PTMs on CAR-T cell functionality is systematically explored in this review, with a particular emphasis on their regulatory roles in key processes such as T cell activation, immune checkpoint modulation, and metabolic reprogramming. Specifically, phosphorylation is crucial in governing T cell activation and exhaustion; ubiquitination is involved in modulating immune checkpoint stability; glycosylation impacts immune synapse formation; and acetylation and lactylation are key in shaping metabolic adaptations crucial for sustained CAR-T efficacy. By leveraging PTM-targeted strategies-including kinase inhibitors, deubiquitinase modulators, and metabolic pathway interventions-CAR-T therapy can be optimized to overcome its current limitations. This review highlights the transformative potential of PTM-based approaches in advancing precision cancer immunotherapy and provides a theoretical foundation for future research and therapeutic innovation.
    Keywords:  CAR-T therapy; Epigenetic reprogramming; Post-translational modifications; T cell fate; Tumor microenvironment modulation
    DOI:  https://doi.org/10.1016/j.critrevonc.2025.104968
  10. Mol Cancer Ther. 2025 Sep 26.
      Metabolic reprogramming constitutes a key mechanism driving immunotherapy resistance in colorectal cancer (CRC), though the immunomodulatory role of L-arginine metabolism remains poorly defined. Through metabolomic profiling, we identified aldehyde dehydrogenase 2 (ALDH2) as a critical regulator depleting intracellular L-arginine pools in CRC cells. High Performance Liquid Chromatography (HPLC) analysis of cell supernatants further demonstrated that ALDH2 overexpression significantly diminishes extracellular L-arginine availability. Functionally, this arginine deficiency suppressed CD8+ T cell proliferation while inducing the attenuation of anti-tumor efficacy. Mechanistic studies revealed that ALDH2 upregulates Pre-B-Cell Leukemia Homeobox 3 (PBX3), which enhances arginase 2 (ARG2) transcription to promote L-arginine catabolism. This process suppresses glycolysis in CD8+ T cells, ultimately compromising their effector functions. Notably, ALDH2-high tumors exhibited resistance to immune checkpoint blockade (ICB), whereas combinatorial ARG2 inhibition and ICB therapy synergistically restored antitumor immunity. These findings nominate ARG2 as a novel therapeutic target and propose dual metabolic-immunologic intervention as a promising strategy for ICB-resistant CRC.
    DOI:  https://doi.org/10.1158/1535-7163.MCT-25-0404
  11. bioRxiv. 2025 Sep 20. pii: 2025.09.17.676943. [Epub ahead of print]
      Dietary components and metabolites play a critical role in regulating intestinal stem and progenitor cell function and proliferation. Here we show that Stearoyl-CoA Desaturases (SCDs), which regulate intracellular saturated to monounsaturated fatty acids ratios, are induced in response to nutrient abundance, especially in the distal intestine, and regulate intestinal homeostasis. Genetic or pharmacological inhibition of SCDs altered lipid metabolism, increased ER stress, and reduced proliferative intestinal stem and progenitor cells in intestinal organoids. These effects were largely mitigated by oleic acid supplementation. Intestinal epithelium-specific deletion of Scd1 and Scd2 led to metabolic rewiring, leading to expansion of progenitor cell populations. DSS-induced epithelial damage revealed a dependence on SCD enzymes during regeneration, accelerating epithelial damage and inflammation in intestines lacking epithelial Scd1 and Scd2 . These findings underscore key metabolic pathways and dependencies that enable intestinal stem and progenitor cells to adapt to nutrient fluctuations and support epithelial tissue regeneration following injury.
    DOI:  https://doi.org/10.1101/2025.09.17.676943
  12. Nature. 2025 Sep 24.
      Chimeric antigen receptor (CAR) T cells are highly effective in haematological malignancies1. However, progressive loss of CAR T cells contributes to relapse in many patients2-4. Here we performed in vivo loss-of-function CRISPR screens in CAR T cells targeting B cell maturation antigen to investigate genes that influence CAR T cell persistence and function in a human multiple myeloma model. We tracked the expansion and persistence of CRISPR library-edited T cells in vitro and at early and late time points in vivo to track the performance of gene-modified CAR T cells from manufacturing to survival in tumours. The screens revealed context-specific regulators of CAR T cell expansion and persistence. Ablation of RASA2 and SOCS1 enhanced T cell expansion in vitro, whereas loss of PTPN2, ZC3H12A and RC3H1 conferred early growth advantages to CAR T cells in vivo. Notably, we identified cyclin-dependent kinase inhibitor 1B (encoded by CDKN1B), a cell cycle regulator, as the most important factor limiting CAR T cell fitness at late time points in vivo. CDKN1B ablation increased CAR T cell proliferation and effector function, significantly enhancing tumour clearance and overall survival. Our findings reveal differing effects of gene perturbation on CAR T cells over time and in different environments, highlight CDKN1B as a promising target to generate highly effective CAR T cells for multiple myeloma and underscore the potential of in vivo screening for identifying genes to enhance CAR T cell efficacy.
    DOI:  https://doi.org/10.1038/s41586-025-09489-8
  13. Cells. 2025 Sep 19. pii: 1467. [Epub ahead of print]14(18):
      The emergence of purinergic signaling has given ATP and other extracellular nucleotides a whole new perspective. This concept refers to the fact that extracellular nucleotides released by most cells act as signaling molecules via specific membrane receptors called purinergic receptors, thus regulating several cellular functions. Although purinergic signaling has been well studied in several physiological systems, recent work has shown that this signaling pathway is also essential in the immune system. In this review, we will discuss the complex role of purinergic signaling in the regulation of effector T cell functions, including migration, differentiation, and activation. We will also cover the role of P2 receptors in the development of autoimmune diseases. Understanding how P2 receptors regulate effector T cells will likely further our understanding of protective immunity and immune pathogenesis and may lead to new therapeutic approaches and agents in immune diseases.
    Keywords:  P2 receptors; autoimmunity; effector T cells; human Th17 cells
    DOI:  https://doi.org/10.3390/cells14181467
  14. bioRxiv. 2025 Sep 15. pii: 2025.09.09.675163. [Epub ahead of print]
      Selenoproteins are involved in immune cell metabolism, yet the roles of these proteins in T cell development and function remain largely unknown. The Trsp gene encodes the selenocysteine tRNA (tRNA Sec ) required for translation of all selenoproteins. In this study, we found that Trsp was required for thymopoiesis, with the majority of tRNA Sec -deficient T cells not progressing beyond double negative 3 stage, with egressed thymocytes undergoing peripheral homeostatic expansion. Trsp- deficient CD4 + T cells exhibited impairments in TCR and IL-2 signaling and did not cause inflammation in experimental models. On the other hand, Trsp -deficient regulatory T (Treg) cells exhibited defects in suppressive function ex vivo and Treg-specific Trsp deletion using Trsp fl/fl Foxp3 YFP-Cre ( Trsp !ιTreg ) mice caused fatal autoimmunity similar to FOXP3-deficient mice. Reducing oxidative stress via 2-HOBA administration prolonged survival in these Trsp !ιTreg mice. These findings indicate that tRNA Sec is required for T cell homeostasis and may be therapeutic targets in inflammation.
    One sentence summary: Trsp , a gene required for translation of all selenoproteins, is essential for all T cell development and function, especially regulatory T cells.
    DOI:  https://doi.org/10.1101/2025.09.09.675163
  15. bioRxiv. 2025 Sep 21. pii: 2025.09.19.677388. [Epub ahead of print]
      Lung adenocarcinoma (LUAD) is the most common form of lung cancer and a leading cause of cancer-related mortality, underscoring the need for new chemopreventive strategies. α-Ketoglutarate (α-KG), a tricarboxylic acid cycle metabolite and dioxygenase cofactor, links cellular metabolism to chromatin regulation. Here, we show that dietary α-KG remodels LUAD in a sex-dependent manner. In female mice, α-KG reduced tumor area, decreased repressive histone marks (H3K27me3, H3K9me3), and upregulated TBX5 and myogenesis-associated genes. In male mice, α-KG-treated male mice exhibited increased tumor area, elevated H3K27me3, and immune remodeling characterized by CD8⁺ T cell expansion and transcriptomic signatures of T cell exhaustion. Analysis of human LUAD revealed that TBX5 expression is enriched in female tumors and associated with improved survival, suggesting it may serve as a marker of favorable outcome. Together, these findings support α-KG as an epigenetic modulator with potential chemopreventive activity in lung cancer and highlight the importance of incorporating sex as a biological variable in preclinical studies.
    DOI:  https://doi.org/10.1101/2025.09.19.677388
  16. Immunity. 2025 Sep 25. pii: S1074-7613(25)00414-5. [Epub ahead of print]
      Metabolic stress in the tumor microenvironment (TME) promotes T cell dysfunction and immune checkpoint inhibitor (ICI) resistance. We examined the contribution of activating transcription factor 4 (ATF4), the central node of the integrated stress response (ISR), to T cell dysfunction in tumors. CD8+ tumor-infiltrating lymphocytes (TILs) in patient samples exhibited chronic ATF4 activity, which was reflected across various tumor models. Hypoxia in the TME imposed chronic ATF4 activity via the ISR kinases. ATF4 overexpression in CD8+ T cells induced metabolic polarity, mitochondrial oxidative stress, and cell death, impairing antitumor immunity. Chronic ATF4 transcriptional activity replicated the terminal exhaustion CD8+ T cell state independent of T cell receptor (TCR) stimulation. Genetic or pharmacologic attenuation of ATF4 reduced mitochondrial oxidative stress and promoted CD8+ TIL viability, enabling response to programmed cell death protein-1 (PD-1) inhibitor therapy and conferring protection from re-emergent disease. Thus, the ISR converges on chronic ATF4 activity in CD8+ TILs as a barrier to ICI response, positioning ISR therapeutics as candidates for immunotherapy.
    Keywords:  ATF4; T cell; hypoxia; immunotherapy; integrated stress response; metabolism; mitochondria; tumor microenvironment
    DOI:  https://doi.org/10.1016/j.immuni.2025.09.003
  17. bioRxiv. 2025 Sep 16. pii: 2025.09.11.675648. [Epub ahead of print]
      Adoptive cell transfer (ACT) of engineered T cells is effective against B-cell malignancies but has faltered against solid tumors due to the immunosuppressive tumor microenvironment (TME). FASL and TGFβ are key mediators of T cell dysfunction in the TME and overexpressing dominant negative (dn) forms of their receptors in T cells increases anti-tumor efficacy in solid tumor models. However, an approach which directly targets the endogenous genes would be more amenable to multiplex editing and reduce competition with WT alleles. Here, we employ base editing (BE) in primary human T cells to install naturally occurring dominant negative FAS and TGFβR2 mutations. In vitro survival and proliferation assays demonstrate that BE T cells are resistant to pro-apoptotic and anti-proliferative effects of FAS and TGFβ signaling. CAR-T cells with BE-installed dn TGFβR2 or dn FAS exhibit improvements in cytotoxicity, while dn TGFβR2 CAR T demonstrate increased persistence and reduced expression of phenotypic markers of exhaustion compared to controls. Moreover, BE-engineered dn CAR T outperform lentiviral-engineered cDNA over expression counterparts in several functional assays. Considering the efficiency of BE and its amenability for multiplex editing, our novel approach lends itself to engineering strategies necessary to overcome T cell dysfunction in solid tumors.
    DOI:  https://doi.org/10.1101/2025.09.11.675648
  18. J Immunol. 2025 Sep 16. pii: vkaf239. [Epub ahead of print]
      The differentiation of CD8 T cells into effector and memory populations is guided by a combination of antigenic, costimulatory, and cytokine signals. Here we show that, within 24 h of activating naïve CD8 T cells, populations emerge with divergent patterns of interleukin (IL)-2 and interferon (IFN)-γ synthesis. This rapid, dynamic, and heterogeneous burst of cytokine production manifests with every CD8 T cell specificity analyzed, is apparent in vivo and in vitro, and occurs prior to the first cell division. Nevertheless, how the intrinsic manufacture of distinct cytokines forecasts and influences the properties and fates of the producer cell itself are not well defined. We demonstrate that the initial cell intrinsic synthesis of IL-2 attenuates IL-2-dependent STAT5 signaling, but that this is not due to differences in the surface expression of the IL-2 receptor complex. The functionally discrete subsets are transcriptionally distinct and display differences in the expression of hallmark effector and memory associated genes. Using cytokine reporter systems, we reveal that these early functional differences are consequential for establishing fate biases and directing the gain of effector and memory T cell properties. The bifurcation between the abilities of IL-2-producing and non-producing subsets to elaborate STAT5 signaling is consistent with a model in which non-IL-2-producing CD8 T cells are more receptive to extrinsic IL-2 signals and preferentially contribute to the early surge of effector formation. Despite this, both IL-2-producing and non-producing CD8 T cells can go on to acquire memory traits, indicating that there is developmental diversity within each cytokine producing subset.
    Keywords:  T cells; cytokines; effector; memory
    DOI:  https://doi.org/10.1093/jimmun/vkaf239
  19. Aging Cell. 2025 Sep 26. e70247
      Mitochondrial calcium (Ca2+) homeostasis plays a critical role in aging and cellular fitness. In the search for novel antiaging approaches, we explored how genetic and pharmacological inhibition of mitochondrial Ca2+ uptake influences the lifespan and health of Caenorhabditis elegans. Using live-cell imaging, we demonstrate that RNA interference-mediated knockdown of mcu-1, the nematode ortholog of the mitochondrial Ca2+ uniporter (MCU), reduces mitochondrial Ca2+ levels, thereby extending lifespan and preserving motility during aging, while compromising early-life survival. This longevity benefit requires intervention before day 14 and coincides with a transient increase in reactive oxygen species (ROS), which activates pathways involving pmk-1, daf-16, and skn-1, orthologs of human p38 mitogen-activated protein kinase (p38 MAPK), forkhead box O (FOXO), and nuclear factor erythroid 2-related factor 2 (NRF2), respectively. This pathway promotes antioxidant defense mechanisms and preserves mitochondrial structure and function during aging, maintaining larger, more interconnected mitochondria and restoring the oxidized/reduced nicotinamide adenine dinucleotide (NAD+/NADH) ratio and oxygen consumption rates to youthful levels. Pharmacological inhibition of mitochondrial Ca2+ uptake using the MCU inhibitor mitoxantrone mirrors the effects of mcu-1 knockdown, extending lifespan and improving fitness in aged nematodes. In human foreskin fibroblasts, short-term mitoxantrone treatment also transiently elevates ROS production and induces enhanced expression and activity of antioxidant defense enzymes, underscoring the translational relevance of findings from nematodes to human cells. Our findings suggest that modulation of mitochondrial Ca2+ uptake induces mitohormesis through ROS-mediated signaling, promoting improved longevity and healthspan in nematodes, with possible implications for healthy aging in humans.
    Keywords:   C. elegans ; aging; lifespan; longevity; mitochondria; reactive oxygen species
    DOI:  https://doi.org/10.1111/acel.70247
  20. Proc Natl Acad Sci U S A. 2025 Sep 30. 122(39): e2419888122
      The five-year survival rate for ovarian cancer patients remains below 50%, underscoring the need for innovative therapies. One promising approach involves engineering T cells to specifically target proteins uniquely overexpressed in tumors, thereby controlling tumor growth without toxicity to healthy tissues. Mesothelin (MSLN) contributes to the malignant and invasive phenotype in ovarian cancer and has limited expression in healthy cells, making it a candidate immunotherapy target. Our previous results in a mouse model of ovarian cancer demonstrated that T cells engineered to express a T cell receptor (TCR) targeting MSLN (TCRMSLN) mediated therapeutic activity, delaying tumor growth and prolonging mouse survival. However, inhibitory ligands expressed in the tumor microenvironment (TME) interacted with inhibitory receptors on activated T cells, suppressing antitumor function. We hypothesized combining engineered T cells with checkpoint blockade would enhance T cell function and improve therapeutic efficacy, but administration of monospecific antibodies targeting individual inhibitory pathways had no significant impact on T cell efficacy. By contrast, the combination of PD-1, Tim-3, and Lag-3 blockade with engineered T cells significantly improved T cell function and overall animal survival relative to treatment with antibody alone or TCRMSLN with singlet or doublet antibody combinations. Single-cell RNA sequencing revealed TCRMSLN T cells treated with the triplet antibody combination increased expression of genes involved in interferon responses and metabolic function, and reduced expression of genes associated with exhaustion. These results suggest that strategies to disrupt multiple inhibitory pathways simultaneously may be necessary for improved adoptive T cell therapy efficacy in patients.
    Keywords:  T cell engineering; adoptive cell therapy; checkpoint blockade; immunotherapy; ovarian cancer
    DOI:  https://doi.org/10.1073/pnas.2419888122
  21. Biochem Pharmacol. 2025 Sep 23. pii: S0006-2952(25)00624-0. [Epub ahead of print]242(Pt 3): 117359
      Aging is an inevitable biological process associated with progressive physiological decline and increased disease susceptibility. Cellular senescence stands as a key mechanism among the hallmarks of aging, which is characterized by irreversible cell-cycle arrest, chromatin remodeling, and a pro-inflammatory senescence-associated secretory phenotype (SASP). Importantly, SASP drives inflammaging and propagates senescence via a bystander effect, exacerbating tissue dysfunction. Recent advances in senolytic therapies and senostatics offer promising strategies to eliminate or rejuvenate senescent cells, improving physiological function in aged and disease models. Notably, panobinostat has emerged as an effective post-chemotherapy senolytic, mitigating chemoresistance. However, current senolytics face challenges, including off-target effects and limited clinical applicability. Growing evidence highlights natural products (e.g., polyphenols, flavonoids) and stem cell therapies as potential anti-aging interventions, with demonstrated efficacy in age-related disease models and ovarian rejuvenation. Despite progress, key hurdles remain in developing personalized, multi-target therapies that safely modulate aging trajectories. This review explores the mechanisms of cellular senescence, anti-aging mechanisms of phytochemicals, and phytochemicals and stem cell-therapy in ovary rejuvenation. We further discuss the challenges in developing "tailor-made" anti-aging interventions that rewire the aging trajectory, which will be critical for achieving healthy aging.
    Keywords:  Anti-aging intervention; Cellular senescence; Chemoresistance; Natural product; Stem cell therapy
    DOI:  https://doi.org/10.1016/j.bcp.2025.117359
  22. Proc Natl Acad Sci U S A. 2025 Sep 30. 122(39): e2506417122
      Prior to ejaculation, mammalian sperm are stored in the epididymis in a "resting" metabolic state. Upon ejaculation, sperm must alter their metabolism to generate the energy needed to support the motility and maturation process known as capacitation to reach and fertilize the oocyte. How sperm regulate the capacitation-induced increase in carbon flux is unknown. Here, we use 13C stable isotope labeling in mouse sperm isolated from the cauda epididymis to follow glucose metabolism through central carbon metabolic network before and after sperm activation. As sperm transition from resting to highly activated states, they boost energy yield by increasing flux through glycolysis at the expense of the pentose phosphate pathway. Increased glycolytic activity seems to be achieved via capacitation-induced stimulation of flux through aldolase. In the mitochondria-containing midpiece, glycolytically generated pyruvate feeds the tricarboxylic acid (TCA) cycle to further maximize energy yield via oxidative phosphorylation. In the mitochondria-free principal piece of the flagellum, pyruvate produced from glycolysis is reduced to lactate by lactate dehydrogenase, which also serves to regenerate oxidized nicotinamide adenine dinucleotide (NAD+) ensuring a sufficient supply to support glycolysis. The resultant lactate is at least partially secreted. Finally, we find evidence that there is an as yet unknown endogenous source of energy in sperm, feeding the upregulation of TCA cycle intermediates. These studies provide the most complete picture of the metabolic shift which occurs in capacitating mouse sperm in glucose.
    Keywords:  aldolase; glycolysis; metabolic reprogramming; sperm; stable isotope labeling
    DOI:  https://doi.org/10.1073/pnas.2506417122
  23. Nature. 2025 Sep 24.
      Chimeric antigen receptor (CAR) T cell therapy has shown remarkable success in treating blood cancers, but CAR T cell dysfunction remains a common cause of treatment failure1. Here we present CELLFIE, a CRISPR screening platform for enhancing CAR T cells across multiple clinical objectives. We performed genome-wide screens in human primary CAR T cells, with readouts capturing key aspects of T cell biology, including proliferation, target cell recognition, activation, apoptosis and fratricide, and exhaustion. Screening hits were prioritized using a new in vivo CROP-seq2 method in a xenograft model of human leukaemia, establishing several gene knockouts that boost CAR T cell efficacy. Most notably, we discovered that RHOG knockout is a potent and unexpected CAR T cell enhancer, both individually and together with FAS knockout, which was validated across multiple in vivo models, CAR designs and sample donors, and in patient-derived cells. Demonstrating the versatility of the CELLFIE platform, we also conducted combinatorial CRISPR screens to identify synergistic gene pairs and saturation base-editing screens to characterize RHOG variants. In summary, we discovered, validated and biologically characterized CRISPR-boosted CAR T cells that outperform standard CAR T cells in widely used benchmarks, establishing a foundational resource for optimizing cell-based immunotherapies.
    DOI:  https://doi.org/10.1038/s41586-025-09507-9
  24. J Immunol. 2025 Sep 22. pii: vkaf196. [Epub ahead of print]
      In response to an infection, the host T cell compartment develops immunological memory to ensure rapid responses upon re-infection with the same pathogen. However, these memory responses can also modulate immune reactions to unrelated pathogens through bystander activation. Herein, T cells are activated in an antigen-independent manner, often triggered by innate cytokines or Toll-like receptor ligands. To uncover new strategies for modulating immune responses, it is essential to deepen our understanding of this alternative mechanism of T cell activation, particularly in humans. Therefore, we studied the response of human CD8+ T cells to innate bystander stimuli in vitro. Thereby, we measured the induction of activation markers and proliferation using flow cytometry, and depleted or blocked several potential modulatory components to identify mediators of bystander CD8+ T-cell responses. Our study demonstrates that CD8+ T cells can be activated as bystander cells in response to innate stimuli present during a viral infection, including IL-15, IFN-α, and TLR7/8 and 9 agonists (R848 and CpG). Depletion experiments demonstrated that these bystander responses are dependent on monocytes and CD4+ T cells. In addition, we revealed that the bystander responses to the innate stimuli are highly reliant on IL-2 signaling. Altogether, our study underscores the pivotal role of IL-2 in mediating bystander responses of CD8+ T cells to innate stimuli, revealing a novel mechanism of immune response modulation during viral infections.
    Keywords:  IL-2; T cells; bystander activation
    DOI:  https://doi.org/10.1093/jimmun/vkaf196
  25. Drug Des Devel Ther. 2025 ;19 8489-8522
       Background: Cellular senescence is a fundamental characteristic of aging, marked by permanent cell cycle cessation and the release of pro-inflammatory mediators. Although senescence plays advantageous roles in tissue regeneration and tumor suppression, its accumulation leads to aging-related illnesses and functional deterioration.
    Objective: This review examines the processes of cellular senescence, its effects on aging and age-related disorders, and emerging therapeutic strategies to modulate senescence for promoting healthy aging.
    Methods: A thorough literature review was performed using peer-reviewed studies on cellular senescence, its molecular pathways, and therapeutic interventions. Emphasis was placed on senolytics, senomorphics, and lifestyle interventions that modulate senescence-associated pathways. Studies published in Scopus, Web of Science and PubMed between 2014-2025 were selected.
    Results: Recent discoveries underscore the dual function of cellular senescence in aging and pathology. The senescence-associated secretory phenotype (SASP) fosters chronic inflammation and tissue dysfunction, connecting senescence to age-related diseases including cardiovascular conditions, dementia, and metabolic disorders. Therapeutic strategies, including senolytics (drugs that specifically eradicate senescent cells) and senomorphics (compounds that suppress SASP without killing cells), show promise in preclinical and clinical studies. Notably, dosing interals (intermittent vs continuous) influence both therapeutic efficacy and adverse events such as thrombocytopenia. Additionally, the state and limitations of clinical validation of aging biomarkers (eg, p16^INK4a, β-galactosidase) remain major hurdles for translation. Lifestyle interventions such as calorie restriction and exercise have also been identified as natural modulators of senescence pathways.
    Conclusion: Targeting cellular senescence offers a promising avenue for promoting healthy aging and mitigating age-linked diseases. Continued research into senescence-modulating interventions may lead to novel therapeutics designed to prolong healthspan and lifespan.
    Keywords:  SASP; age-related diseases; aging; cellular senescence; healthy aging; senolytics; senomorphics
    DOI:  https://doi.org/10.2147/DDDT.S543211
  26. JID Innov. 2025 Nov;5(6): 100403
      Circulating cutaneous lymphocyte-associated antigen-positive T cells constitute a subset of memory T cells with a unique phenotype, effector function, and therapeutic relevance because they reflect the regional functions of the cutaneous immune system. These cells are involved in the pathological mechanisms of diverse cutaneous diseases. This review brings updated translational insights into these cells and identifies key questions for future research in the field.
    Keywords:  CLA+ T cells; Immunodermatology; Skin diseases; Translational
    DOI:  https://doi.org/10.1016/j.xjidi.2025.100403
  27. Sci Signal. 2025 Sep 23. 18(905): eadr3177
      Metabolic reprogramming controls protective and pathogenic T helper 17 (TH17) cell responses. When naïve T cells are differentiated into TH17 cells in vitro, the presence of the cytokine activin A promotes their maturation into a nonpathogenic state. Here, we found that nonpathogenic TH17 cells induced by activin A displayed reduced aerobic glycolysis and increased oxidative phosphorylation (OXPHOS). In response to activin A, signaling through the adenosine A2A receptor (A2AR) and AMP-activated protein kinase (AMPK) enhanced OXPHOS and reprogrammed pathogenic TH17 cells toward nonpathogenic states that did not induce central nervous system autoimmunity in a mouse model of multiple sclerosis. In pathogenic TH17 cells, the transcriptional coactivator p300/CBP-associated factor (PCAF) increased acetylation at histone 3 Lys9 (H3K9ac) of genes involved in aerobic glycolysis and TH17 pathogenic programs. In contrast, in nonpathogenic activin A-treated TH17 cells, AMPK signaling suppressed PCAF-mediated H3K9ac modification of genes involved in aerobic metabolism and enhanced H3K9ac modification of genes involved in OXPHOS and nonpathogenic TH17 programs. Together, our findings uncover A2AR-AMPK signaling as a central metabolic checkpoint that suppresses TH17 cell pathogenicity.
    DOI:  https://doi.org/10.1126/scisignal.adr3177
  28. Brain Commun. 2025 ;7(5): fcaf330
      It is known that T cells play an important role in the progression of neurodegenerative disorders, including tauopathies. In this study, we used fingolimod (FTY720), an approved medication for the treatment of multiple sclerosis (MS), to manipulate T cell dynamics in P301S-Tau transgenic (Tau Tg) mice. FTY720 dramatically decreased the population of circulating T cells in blood. However, unexpectedly, we observed a marked increase in the number of CD8+ T cells in the hippocampus of FTY720-treated Tau Tg mice. This increase in CD8+ T cell number was significantly correlated with enhanced tau phosphorylation. Notably, FTY720-treated Tau Tg mice exhibited brain atrophy and neurodegeneration compared with controls. These findings indicate that CD8+ T cells in the brain contribute to the progression of tauopathies, and that brain CD8+ T cells may be a promising target for the treatment of tauopathies. This study provides new insights into the dynamics of brain T cells in neurodegenerative disorders. In addition, these results raise caution regarding FTY720 treatment in individuals predisposed to tauopathies, as it may promote neurodegeneration despite reducing peripheral T cells.
    Keywords:  FTY720; Neurodegeneration; Phosphorylated tau; T cell; Tauopathy
    DOI:  https://doi.org/10.1093/braincomms/fcaf330
  29. J Adv Res. 2025 Sep 18. pii: S2090-1232(25)00729-5. [Epub ahead of print]
       BACKGROUND: Tumor-associated macrophages (TAMs) depend on their amino acid metabolism to determine their properties and immune function and play important roles in the tumor microenvironment (TME). Although in previous studies, targeting amino acid metabolism to transform the protumor function of TAMs into antitumor immune function has shown promising application as a tumor therapy, current clinical research is still limited. There is a lack of discussion on the mechanism and treatment strategy for determining tumor progression by controlling amino acid metabolism in TAMs, as does a summary of studies on promoting tumor progression by reshaping amino acid metabolism in TAMs.
    AIM OF REVIEW: This review aims to systematically review and summarize the crosstalk between amino acid metabolism in TAMs and the TME, analyze the determining role of its metabolic network in tumor occurrence and development, and summarize therapies on this basis to help determine the development status and emerging technologies in the field of amino acid metabolism in TAMs for tumor therapy.
    KEY SCIENTIFIC CONCEPTS OF REVIEW: This review dissects how TAMs exploit amino acid dynamics via transporters, enzymes, and sensors to adopt protumoral phenotypes, depleting critical metabolites and crippling antitumor T-cell responses. We map the immunometabolic crosstalk through which TAMs reshape immunity, highlighting nutrient competition and metabolic byproducts as dual drivers of immune dysfunction. Emerging therapeutic strategies targeting these pathways (IFN-γ-JAK-STAT1 and IL-6/JAK2/STAT3) have been critically evaluated for their potential to reprogram TAMs and reverse immunosuppression. Key challenges, such as TAM heterogeneity, metabolic plasticity, and therapy resistance, are addressed, emphasizing the need for single-cell-resolution mapping of TAM metabolic states to identify context-dependent vulnerabilities. Finally, we advocate for combinatorial approaches that couple metabolic rewiring with immunotherapies, proposing that disrupting amino acid dependencies in TAMs could dismantle the immunosuppressive TME.
    Keywords:  Amino acid; Immunometabolic crosstalk; Immunotherapy; M1-like TAMs; M2-like TAMs; Metabolic crosstalk; Metabolic reprogramming; Therapeutic target; Tumor microenvironment (TME); Tumor-associated macrophages (TAMs)
    DOI:  https://doi.org/10.1016/j.jare.2025.09.025